151
|
D’Silva SZ, Singh M, Pinto AS. NK cell defects: implication in acute myeloid leukemia. Front Immunol 2023; 14:1112059. [PMID: 37228595 PMCID: PMC10203541 DOI: 10.3389/fimmu.2023.1112059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is a complex disease with rapid progression and poor/unsatisfactory outcomes. In the past few years, the focus has been on developing newer therapies for AML; however, relapse remains a significant problem. Natural Killer cells have strong anti-tumor potential against AML. This NK-mediated cytotoxicity is often restricted by cellular defects caused by disease-associated mechanisms, which can lead to disease progression. A stark feature of AML is the low/no expression of the cognate HLA ligands for the activating KIR receptors, due to which these tumor cells evade NK-mediated lysis. Recently, different Natural Killer cell therapies have been implicated in treating AML, such as the adoptive NK cell transfer, Chimeric antigen receptor-modified NK (CAR-NK) cell therapy, antibodies, cytokine, and drug treatment. However, the data available is scarce, and the outcomes vary between different transplant settings and different types of leukemia. Moreover, remission achieved by some of these therapies is only for a short time. In this mini-review, we will discuss the role of NK cell defects in AML progression, particularly the expression of different cell surface markers, the available NK cell therapies, and the results from various preclinical and clinical trials.
Collapse
Affiliation(s)
- Selma Z. D’Silva
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Meenakshi Singh
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Andrea S. Pinto
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| |
Collapse
|
152
|
Li X, Wang HY, Gao F, Guo FF, Wang XN, Pan YX, Bai GQ. Tenofovir alters the immune microenvironment of pregnant women with hepatitis B virus infection: Evidence from single-cell RNA sequencing. Int Immunopharmacol 2023; 119:110245. [PMID: 37163920 DOI: 10.1016/j.intimp.2023.110245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/16/2023] [Accepted: 04/23/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Mother-to-child is the main route of the transmission of hepatitis B virus (HBV) infection. Tenofovir fumarate (TDF) antiviral treatment has become the most extensive choice worldwide. However, the effects of TDF treatment on the immune function of pregnant women remains unclear. Here we investigate the effect of TDF treatment on the immune microenvironment of pregnant women with HBV infection using single-cell RNA sequencing (scRNA-seq). METHODS Three HBV-infected pregnant women were treated with TDF and six samples were collected before and after the treatment. In total, 68,200 peripheral blood mononuclear cells (PBMCs) were extracted for 10 × scRNA-seq. The cells were clustered using t-distributed stochastic neighbor embedding (t-SNE) and unbiased computational informatics analysis. RESULTS The analysis identified four-cell subtypes, including T cells, monocytes, natural killer (NK) cells, and B cells, and unraveled the developmental trajectory and maturation of CD4+ T and CD8+ T cell subtypes. The cellular state and molecular features of the effector/memory T cells revealed a significant increase in the inflammatory state of CD4+ T cells and the cytotoxic characteristics of CD8+ T cells. Additionally, after TDF treatment, the monocytes showed a tendency for M1 polarization, and the cytotoxicity of NK cells was enhanced. Furthermore, the analysis of intercellular communication revealed the interaction of various subtypes of cells and the heterogeneous expression of key signal pathways. CONCLUSIONS The findings of this study reveal significant differences in cellular subtypes and molecular characteristics of PBMCs of pregnant women with HBV infection before and after TDF treatment and demonstrate the recovery of immune response after treatment. These findings could help develop immune intervention measures to control HBV during pregnancy and the puerperium period.
Collapse
Affiliation(s)
- Xia Li
- Gene Joint Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hong-Yan Wang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fan Gao
- Clinical Research Center, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fan-Fan Guo
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiao-Na Wang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yi-Xia Pan
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Gui-Qin Bai
- Gene Joint Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
153
|
Duane C, O'Dwyer M, Glavey S. Adoptive Immunotherapy and High-Risk Myeloma. Cancers (Basel) 2023; 15:cancers15092633. [PMID: 37174099 PMCID: PMC10177276 DOI: 10.3390/cancers15092633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Despite significant improvements in the treatment of multiple myeloma (MM), it remains mostly incurable, highlighting a need for new therapeutic approaches. Patients with high-risk disease characteristics have a particularly poor prognosis and limited response to current frontline therapies. The recent development of immunotherapeutic strategies, particularly T cell-based agents have changed the treatment landscape for patients with relapsed and refractory disease. Adoptive cellular therapies include chimeric antigen receptor (CAR) T cells, which have emerged as a highly promising therapy, particularly for patients with refractory disease. Other adoptive cellular approaches currently in trials include T cell receptor-based therapy (TCR), and the expansion of CAR technology to natural killer (NK) cells. In this review we explore the emerging therapeutic field of adoptive cellular therapy for MM, with a particular focus on the clinical impact of these therapies for patients with high-risk myeloma.
Collapse
Affiliation(s)
- Catherine Duane
- Department of Haematology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
| | - Michael O'Dwyer
- Department of Haematology, University of Galway, H91 TK33 Galway, Ireland
| | - Siobhan Glavey
- Department of Haematology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
- Department of Pathology, Royal College of Surgeons in Ireland, D09 V2N0 Dublin, Ireland
| |
Collapse
|
154
|
Cimpean M, Cooper MA. Metabolic regulation of NK cell antiviral functions during cytomegalovirus infection. J Leukoc Biol 2023; 113:525-534. [PMID: 36843434 PMCID: PMC11262056 DOI: 10.1093/jleuko/qiad018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/28/2023] Open
Abstract
Natural killer (NK) cells quickly mount cytotoxic responses, produce cytokines, and proliferate in response to infected or transformed cells. Moreover, they can develop memory, with enhanced effector responses following activation, in some cases with antigen specificity. To optimally execute these functions, NK cells undergo metabolic reprogramming. Here, we discuss the interplay between metabolism and NK cell function in the context of viral infections. We review findings supporting metabolic regulation of NK cell effector functions, with a focus on NK cell antiviral infection in the context of cytomegalovirus in the mouse (MCMV) and human (HCMV).
Collapse
Affiliation(s)
- Maria Cimpean
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, USA
| | - Megan A. Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, USA
| |
Collapse
|
155
|
Roato I, Mauceri R, Notaro V, Genova T, Fusco V, Mussano F. Immune Dysfunction in Medication-Related Osteonecrosis of the Jaw. Int J Mol Sci 2023; 24:ijms24097948. [PMID: 37175652 PMCID: PMC10177780 DOI: 10.3390/ijms24097948] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The pathogenesis of medication-related osteonecrosis of the jaw (MRONJ) is multifactorial and there is a substantial consensus on the role of antiresorptive drugs (ARDs), including bisphosphonates (BPs) and denosumab (Dmab), as one of the main determinants. The time exposure, cumulative dose and administration intensity of these drugs are critical parameters to be considered in the treatment of patients, as cancer patients show the highest incidence of MRONJ. BPs and Dmab have distinct mechanisms of action on bone, but they also exert different effects on immune subsets which interact with bone cells, thus contributing to the onset of MRONJ. Here, we summarized the main effects of ARDs on the different immune cell subsets, which consequently affect bone cells, particularly osteoclasts and osteoblasts. Data from animal models and MRONJ patients showed a deep interference of ARDs in modulating immune cells, even though a large part of the literature concerns the effects of BPs and there is a lack of data on Dmab, demonstrating the need to further studies.
Collapse
Affiliation(s)
- Ilaria Roato
- CIR-Dental School, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Rodolfo Mauceri
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90133 Palermo, Italy
| | - Vincenzo Notaro
- CIR-Dental School, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Tullio Genova
- Department of Life Sciences and Systems Biology, University of Torino, 10123 Torino, Italy
| | - Vittorio Fusco
- Medical Oncology Unit, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
- Department of Integrated Research Activity and Innovation (DAIRI), Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Federico Mussano
- CIR-Dental School, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| |
Collapse
|
156
|
Cimpean M, Keppel MP, Gainullina A, Fan C, Schedler NC, Swain A, Kolicheski A, Shapiro H, Young HA, Wang T, Artyomov MN, Cooper MA. IL-15 priming alters IFN-γ regulation in murine NK cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.23.537947. [PMID: 37163083 PMCID: PMC10168240 DOI: 10.1101/2023.04.23.537947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Natural killer (NK) effector functions can be triggered by inflammatory cytokines and engagement of activating receptors. NK cell production of IFN-γ, an important immunoregulatory cytokine, exhibits activation-specific IFN-γ regulation. Resting murine NK cells exhibit activation-specific metabolic requirements for IFN-γ production, which are reversed for activating receptor-mediated stimulation following IL-15 priming. While both cytokine and activating receptor stimulation leads to similar IFN-γ protein production, only cytokine stimulation upregulates Ifng transcript, suggesting that protein production is translationally regulated after receptor stimulation. Based on these differences in IFN-γ regulation, we hypothesized that ex vivo IL-15 priming of murine NK cells allows a switch to IFN-γ transcription upon activating receptor engagement. Transcriptional analysis of primed NK cells compared to naïve cells or cells cultured with low-dose IL-15 demonstrated that primed cells strongly upregulated Ifng transcript following activating receptor stimulation. This was not due to chromatin accessibility changes in the Ifng locus or changes in ITAM signaling, but was associated with a distinct transcriptional signature induced by ITAM stimulation of primed compared to naïve NK cells. Transcriptional analyses identified a common signature of c-Myc (Myc) targets associated with Ifng transcription. While Myc marked NK cells capable of Ifng transcription, Myc itself was not required for Ifng transcription using a genetic model of Myc deletion. This work highlights altered regulatory networks in IL-15 primed cells, resulting in distinct gene expression patterns and IFN-γ regulation in response to activating receptor stimulation.
Collapse
|
157
|
Yu Y. The Function of NK Cells in Tumor Metastasis and NK Cell-Based Immunotherapy. Cancers (Basel) 2023; 15:cancers15082323. [PMID: 37190251 DOI: 10.3390/cancers15082323] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic tumors cause the most deaths in cancer patients. Treating metastasis remains the primary goal of current cancer research. Although the immune system prevents and kills the tumor cells, the function of the immune system in metastatic cancer has been unappreciated for decades because tumors are able to develop complex signaling pathways to suppress immune responses, leading them to escape detection and elimination. Studies showed NK cell-based therapies have many advantages and promise for fighting metastatic cancers. We here review the function of the immune system in tumor progression, specifically focusing on the ability of NK cells in antimetastasis, how metastatic tumors escape the NK cell attack, as well as the recent development of effective antimetastatic immunotherapies.
Collapse
Affiliation(s)
- Yanlin Yu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
158
|
Althaus J, Nilius-Eliliwi V, Maghnouj A, Döring S, Schroers R, Hudecek M, Hahn SA, Mika T. Cytotoxicity of CD19-CAR-NK92 cells is primarily mediated via perforin/granzyme pathway. Cancer Immunol Immunother 2023:10.1007/s00262-023-03443-1. [PMID: 37052701 PMCID: PMC10361870 DOI: 10.1007/s00262-023-03443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/28/2023] [Indexed: 04/14/2023]
Abstract
Chimeric antigen receptors (CARs) have improved cancer immunotherapy in recent years. Immune cells, such as Natural killer cells (NK-cells) or T cells, are used as effector cells in CAR-therapy. NK92-cells, a cell line with known cytotoxic activity, are of particular interest in CAR-therapy since culturing conditions are simple and anti-tumor efficacy combined with a manageable safety profile was proven in clinical trials. The major pathways of immune effector cells, including NK92-cells, to mediate cytotoxicity, are the perforin/granzyme and the death-receptor pathway. Detailed knowledge of CAR-effector cells' cytotoxic mechanisms is essential to unravel resistance mechanisms, which potentially arise by resistance against apoptosis-inducing signaling. Since mutations in apoptosis pathways are frequent in lymphoma, the impact on CAR-mediated cytotoxicity is of clinical interest. In this study, knockout models of CD19-CAR-NK92 cells were designed, to investigate cytotoxic pathways in vitro. Knockout of perforin 1 (Prf1) and subsequent abrogation of the perforin/granzyme pathway dramatically reduced the cytotoxicity of CD19-CAR-NK92 cells. In contrast, knockout of FasL and inhibition of TRAIL (tumor necrosis factor-related apoptosis-inducing ligands) did not impair cytotoxicity in most conditions. In conclusion, these results indicate the perforin/granzyme pathway as the major pathway to mediate cytotoxicity in CD19-CAR-NK92 cells.
Collapse
Affiliation(s)
- Jonas Althaus
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Verena Nilius-Eliliwi
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, In der Schornau 23-25, D-44892, Bochum, Germany
| | - Abdelouahid Maghnouj
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Sascha Döring
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, In der Schornau 23-25, D-44892, Bochum, Germany
| | - Michael Hudecek
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Stephan A Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Thomas Mika
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany.
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, In der Schornau 23-25, D-44892, Bochum, Germany.
| |
Collapse
|
159
|
Dai X, Zhou X, Shao R, Zhao R, Yanamandra AK, Xing Z, Ding M, Wang J, Liu T, Zheng Q, Zhang P, Zhang H, Wang Y, Qu B, Wang Y. Bioactive Constituents of Verbena officinalis Alleviate Inflammation and Enhance Killing Efficiency of Natural Killer Cells. Int J Mol Sci 2023; 24:ijms24087144. [PMID: 37108306 PMCID: PMC10138337 DOI: 10.3390/ijms24087144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/28/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Natural killer (NK) cells play key roles in eliminating pathogen-infected cells. Verbena officinalis (V. officinalis) has been used as a medical plant in traditional and modern medicine for its anti-tumor and anti-inflammatory activities, but its effects on immune responses remain largely elusive. This study aimed to investigate the potential of V. officinalis extract (VO extract) to regulate inflammation and NK cell functions. We examined the effects of VO extract on lung injury in a mouse model of influenza virus infection. We also investigated the impact of five bioactive components of VO extract on NK killing functions using primary human NK cells. Our results showed that oral administration of VO extract reduced lung injury, promoted the maturation and activation of NK cells in the lung, and decreased the levels of inflammatory cytokines (IL-6, TNF-α and IL-1β) in the serum. Among five bioactive components of VO extract, Verbenalin significantly enhanced NK killing efficiency in vitro, as determined by real-time killing assays based on plate-reader or high-content live-cell imaging in 3D using primary human NK cells. Further investigation showed that treatment of Verbenalin accelerated the killing process by reducing the contact time of NK cells with their target cells without affecting NK cell proliferation, expression of cytotoxic proteins, or lytic granule degranulation. Together, our findings suggest that VO extract has a satisfactory anti-inflammatory effect against viral infection in vivo, and regulates the activation, maturation, and killing functions of NK cells. Verbenalin from V. officinalis enhances NK killing efficiency, suggesting its potential as a promising therapeutic to fight viral infection.
Collapse
Affiliation(s)
- Xiangdong Dai
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiangda Zhou
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Rui Shao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Renping Zhao
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Archana K Yanamandra
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
- Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Zhimei Xing
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mingyu Ding
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Junhong Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tao Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qi Zheng
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Peng Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bin Qu
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Yu Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
160
|
Huang J, Zheng T, Liang Y, Qin Y, Wu X, Fan X. Transcriptome Analysis of Natural Killer Cells in Response to Newcastle Disease Virus Infected Hepatocellular Carcinoma Cells. Genes (Basel) 2023; 14:genes14040888. [PMID: 37107646 PMCID: PMC10138298 DOI: 10.3390/genes14040888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
When tumor cells are infected by the Newcastle disease virus (NDV), the lysis of tumor cells by natural killer (NK) cells is enhanced, which may be related to the enhanced NK cell activation effect. To better understand the intracellular molecular mechanisms involved in NK cell activation, the transcriptome profiles of NK cells stimulated by NDV-infected hepatocellular carcinoma (HCC) cells (NDV group) and control (NC group, NK cells stimulated by HCC cells) were analyzed. In total, we identified 1568 differentially expressed genes (DEGs) in the NK cells of the NDV group compared to the control, including 1389 upregulated and 179 downregulated genes. Functional analysis showed that DEGs were enriched in the immune system, signal transmission, cell growth, cell death, and cancer pathways. Notably, 9 genes from the IFN family were specifically increased in NK cells upon NDV infection and identified as potential prognosis markers for patients with HCC. A qRT-PCR experiment was used to confirm the differential expression of IFNG and the other 8 important genes. The results of this study will improve our understanding of the molecular mechanisms of NK cell activation.
Collapse
Affiliation(s)
- Juanjuan Huang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
- Department of Microbiology, School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Tingting Zheng
- Department of Microbiology, School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Ying Liang
- Department of Microbiology, School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Basic Research on Regional Disease, Education Department of Guangxi, Guangxi Medical University, Nanning 530021, China
| | - Ying Qin
- Department of Microbiology, School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Xing Wu
- Department of Microbiology, School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Xiaohui Fan
- Department of Microbiology, School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Basic Research on Regional Disease, Education Department of Guangxi, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
161
|
Palamarchuk AI, Kovalenko EI, Streltsova MA. Multiple Actions of Telomerase Reverse Transcriptase in Cell Death Regulation. Biomedicines 2023; 11:biomedicines11041091. [PMID: 37189709 DOI: 10.3390/biomedicines11041091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Telomerase reverse transcriptase (TERT), a core part of telomerase, has been known for a long time only for its telomere lengthening function by reverse transcription of RNA template. Currently, TERT is considered as an intriguing link between multiple signaling pathways. The diverse intracellular localization of TERT corresponds to a wide range of functional activities. In addition to the canonical function of protecting chromosome ends, TERT by itself or as a part of the telomerase complex participates in cell stress responses, gene regulation and mitochondria functioning. Upregulation of TERT expression and increased telomerase activity in cancer and somatic cells relate to improved survival and persistence of such cells. In this review, we summarize the data for a comprehensive understanding of the role of TERT in cell death regulation, with a focus on the interaction of TERT with signaling pathways involved in cell survival and stress response.
Collapse
Affiliation(s)
- Anastasia I. Palamarchuk
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Elena I. Kovalenko
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Maria A. Streltsova
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
162
|
Casari G, Dall'Ora M, Melandri A, Masciale V, Chiavelli C, Prapa M, Neri G, Spano MC, Murgia A, D'Esposito A, Baschieri MC, Ceccherelli GB, Dominici M, Grisendi G. Impact of soluble tumor necrosis factor-related apoptosis-inducing ligand released by engineered adipose mesenchymal stromal cells on white blood cells. Cytotherapy 2023; 25:605-614. [PMID: 37012089 DOI: 10.1016/j.jcyt.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/26/2023] [Accepted: 02/15/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND AIMS The proapoptotic protein tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is physiologically expressed by immune cells and performs regulatory functions in infections, autoimmune diseases and cancer, where it acts as a tumor suppressor. Adipose-derived mesenchymal stromal cells (AD-MSCs) also may play immunomodulatory roles in both primary and acquired immune responses. We have previously demonstrated the efficacy of an anticancer gene therapy based on AD-MSC engineered to secrete a soluble TRAIL variant (sTRAIL) against pancreatic cancer. However, the impact of AD-MSC sTRAIL on leukocyte subsets has been not yet considered also to predict a possible immunotoxicity profile in the clinical translation of this cell-based anticancer strategy. METHODS Monocytes, polymorphonuclear cells and T lymphocytes were freshly isolated from the peripheral blood of healthy donors. Immunophenotype and functional (DR4 and DR5) and decoy (DcR1 and DcR2) TRAIL receptors were tested by flow cytometry. The viability of white blood cells treated with sTRAIL released by gene-modified AD-MSC or co-cultured with AD-MSC sTRAIL was then evaluated by both metabolic assays and flow cytometry. In addition, cytokine profile in co-cultures was analyzed by multiplex enzyme-linked immunosorbent assay. RESULTS Monocytes and polymorphonuclear cells showed high positivity for DR5 and DcR2, respectively, whereas T cells revealed negligible expression of all TRAIL receptors. Irrespective of TRAIL receptors' presence on the cell membrane, white blood cells were refractory to the proapoptotic effect displayed by sTRAIL secreted by gene-modified AD-MSC, and direct cell-to-cell contact with AD-MSC sTRAIL had negligible impact on T-cell and monocyte viability. Cytokine crosstalk involving interleukin 10, tumor necrosis factor alpha, and interferon gamma secreted by T lymphocytes and vascular endothelial growth factor A and interleukin 6 released by AD-MSC was highlighted in T-cell and AD-MSC sTRAIL co-cultures. CONCLUSIONS In summary, this study demonstrates the immunological safety and thus the clinical feasibility of an anticancer approach based on AD-MSC expressing the proapoptotic molecule sTRAIL.
Collapse
Affiliation(s)
- Giulia Casari
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy; Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Polytechnic University of Marche, Ancona, Italy
| | | | - Aurora Melandri
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Chiavelli
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Malvina Prapa
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy; Department of Medical Technical Sciences, Universiteti Barleti, Tirana, Albania
| | - Giovanni Neri
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Alba Murgia
- Technopole of Mirandola TPM, Mirandola, Modena, Italy
| | - Angela D'Esposito
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Maria Cristina Baschieri
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | - Massimo Dominici
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy; EVOTEC (Modena) Srl, Medolla, Modena, Italy.
| | - Giulia Grisendi
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
163
|
Arslan M. Whole-genome sequencing and genomic analysis of Norduz goat (Capra hircus). Mamm Genome 2023:10.1007/s00335-023-09990-3. [PMID: 37004528 DOI: 10.1007/s00335-023-09990-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
Artificial and natural selective breeding of goats has resulted in many different goat breeds all around the world. Norduz goat is one of these breeds, and it is a local goat breed of Turkey. The goats are favorable due to pre-weaning viability and reproduction values compared to the regional breeds. Development in sequencing technologies has let to understand huge genomic structures and complex phenotypes. Until now, such a comprehensive study has not been carried out to understand the genomic structure of the Norduz goats, yet. In the study, the next-generation sequencing was carried out to understand the genomic structure of Norduz goat. Real-time PCR was used to evaluate prominent CNVs in the Norduz goat individuals. Whole genome of the goat was constructed with an average of 33.1X coverage level. In the stringent filtering condition, 9,757,980 SNPs, 1,536,715 InDels, and 290 CNVs were detected in the Norduz goat genome. Functional analysis of high-impact SNP variations showed that the classical complement activation biological process was affected significantly in the goat. CNVs in the goat genome were found in genes related to defense against viruses, immune response, and cell membrane transporters. It was shown that GBP2, GBP5, and mammalian ortholog GBP1, which are INF-stimulated GTPases, were found to be high copy numbers in the goats. To conclude, genetic variations mainly in immunological response processes suggest that Norduz goat is an immunologically improved goat breed and natural selection could take an important role in the genetical improvements of the goats.
Collapse
Affiliation(s)
- Mevlüt Arslan
- Department of Genetics, Faculty of Veterinary Medicine, Van Yüzüncü Yıl University, Tuşba, 65080, Van, Turkey.
| |
Collapse
|
164
|
Engineering CAR-NK cells targeting CD33 with concomitant extracellular secretion of anti-CD16 antibody revealed superior antitumor effects toward myeloid leukemia. Cancer Lett 2023; 558:216103. [PMID: 36805460 DOI: 10.1016/j.canlet.2023.216103] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Acute myeloid leukemia (AML) is a common form of acute leukemia, and the currently available treatments are unsatisfactory. In the present study, we report an immune cell therapeutic strategy that employed genetically modified bifunctional CAR-NK cells. These cells combined the efficient targeting of AML cells by the CD33 molecule with the concomitant stimulation of NK cell-mediated cytotoxicity via the expression and extracellular secretion of anti-CD16 antibody (B16) that binds back to the FC receptor of NK cells. Compared to CAR-NK cells that target CD33 only, the bifunctional CD33/B16 CAR-NK cells showed superior killing efficiency toward AML cells in vitro. The increase in efficiency was approximately four-fold, as determined based on the number of cells needed to achieve 80% killing activity. An in vivo study using a xenograft model also revealed the effective clearance of leukemic cells and much longer survival, with no relapse or death for at least 60 days. In addition, the safety of CAR-NK cells did not change with additional expression of B16, as determined by the release of cytokines. These data revealed the development of a promising CAR-NK approach for the treatment of patients with AML, which may improve CAR-NK-based treatment strategy in general and may potentially be used to treat other tumors as well.
Collapse
|
165
|
Wang C, Liu J, Li W. 'Off the shelf' immunotherapies: Generation and application of pluripotent stem cell-derived immune cells. Cell Prolif 2023; 56:e13425. [PMID: 36855955 PMCID: PMC10068955 DOI: 10.1111/cpr.13425] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/10/2023] [Accepted: 01/27/2023] [Indexed: 03/02/2023] Open
Abstract
In recent years, great strides have been made toward the development of immune cell-based therapies in the treatment of refractory malignancies. Primary T cells and NK cells armed with chimeric antigen receptors have achieved tremendous clinical success especially in patients with leukaemia and lymphoma. However, the autologous origin of these effector cells means that a single batch of laboriously engineered cells treats only a certain patient, leading to high cost, ununiform product quality, and risk of delay in treatment, and therefore results in restricted accessibility of these therapies to the overwhelming majority of the patients. Addressing these tricky obstacles calls for the development of universal immune cell products that can be provided 'off the shelf' in a large amount. Pluripotent stem cells (PSCs), owing to their unique capacity of self-renewal and the potential of multi-lineage differentiation, offer an unlimited cell source to generate uniform and scalable engineered immune cells. This review discusses the major advances in the development of PSC-derived immune cell differentiation approaches and their therapeutic potential in treating both hematologic malignancies and solid tumours. We also consider the potency of PSC-derived immune cells as an alternative therapeutic strategy for other diseases, such as autoimmune diseases, fibrosis, infections, et al.
Collapse
Affiliation(s)
- Chenxin Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Bejing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jingjing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Bejing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Bejing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
166
|
Characterization of BV6-Induced Sensitization to the NK Cell Killing of Pediatric Rhabdomyosarcoma Spheroids. Cells 2023; 12:cells12060906. [PMID: 36980247 PMCID: PMC10047333 DOI: 10.3390/cells12060906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Although the overall survival in pediatric rhabdomyosarcoma (RMS) has increased over the last decades, the most aggressive subtype of alveolar RMS is in dire need of novel treatment strategies. RMS cells evade cell death induction and immune control by increasing the expression of inhibitors of apoptosis proteins (IAPs), which can be exploited and targeted with stimulation with Smac mimetics. Here, we used the Smac mimetic BV6 to re-sensitize RMS spheroids to cell death, which increased killing induced by natural killer (NK) cells. Single BV6 treatment of RMS spheroids did not reduce spheroidal growth. However, we observed significant spheroidal decomposition upon BV6 pre-treatment combined with NK cell co-cultivation. Molecularly, IAPs s are rapidly degraded by BV6, which activates NF-κB signal transduction pathways in RMS spheroids. RNA sequencing analysis validated NF-κB activation and identified a plethora of BV6-regulated genes. Additionally, BV6 released caspases from IAP-mediated inhibition. Here, caspase-8 might play a major role, as knockdown experiments resulted in decreased NK cell-mediated attack. Taken together, we improved the understanding of the BV6 mechanism of RMS spheroid sensitization to cytotoxic immune cells, which could be suitable for the development of novel combinatory cellular immunotherapy with Smac mimetics.
Collapse
|
167
|
Higuchi T, Hashida Y, Matsuo K, Kitahata K, Ujihara T, Murakami I, Nakayama T, Daibata M. EBV-positive pyothorax-associated lymphoma expresses CXCL9 and CXCL10 chemokines that attract cytotoxic lymphocytes via CXCR3. Cancer Sci 2023. [PMID: 36898851 DOI: 10.1111/cas.15782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Epstein-Barr virus (EBV)-positive diffuse large B-cell lymphoma associated with chronic inflammation (DLBCL-CI) develops in the setting of long-standing inflammation. This type of lymphoma may have specific expression profiles of chemokines involved in the pathogenesis of DLBCL-CI. EBV-positive pyothorax-associated lymphoma (PAL) is a prototype of DLBCL-CI and represents a valuable model for the study of this disease category. Using a panel of PAL cell lines, we found that PAL cells expressed and secreted C-X-C motif chemokine ligands 9 and 10 (CXCL9 and CXCL10), the ligands of CXCR3, in contrast to EBV-negative DLBCL cell lines, which did not. Culture supernatants from PAL cell lines attracted CXCR3-expressing CD4+ T cells, CD8+ T cells, and CD56+ natural killer cells from human peripheral blood mononuclear cells. PAL cells injected into mice also attracted CXCR3-positive cytotoxic lymphocytes that expressed interferon-γ. The expression of CXCL9 and CXCL10 was detected in PAL tumor biopsy samples from patients, and CXCR3-positive lymphocytes were abundant in the tissue samples. Collectively, these findings suggest that CXCL9 and CXCL10 are produced by PAL cells and can elicit cytotoxic responses via CXCR3. This chemokine system is also likely to contribute to tissue necrosis, which is a signature histological feature of DLBCL-CI. Further studies are warranted to determine whether the CXCL9-CXCL10/CXCR3 axis exerts antitumor effects in DLBCL-CI.
Collapse
Affiliation(s)
- Tomonori Higuchi
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Yumiko Hashida
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Kazuhiko Matsuo
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Japan
| | - Kosuke Kitahata
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Japan
| | - Takako Ujihara
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Nankoku, Japan
- Science Research Center, Kochi University, Nankoku, Japan
| | - Ichiro Murakami
- Department of Pathology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Takashi Nakayama
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Japan
| | - Masanori Daibata
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Nankoku, Japan
| |
Collapse
|
168
|
Gao Y, Zhang X, Jiang T, Zhou H, Liu H, Hu Y, Cao J. Inhibition of hepatic natural killer cell function via the TIGIT receptor in schistosomiasis-induced liver fibrosis. PLoS Pathog 2023; 19:e1011242. [PMID: 36930687 PMCID: PMC10022799 DOI: 10.1371/journal.ppat.1011242] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Schistosomiasis is a zoonotic parasitic disease. Schistosoma japonicum eggs deposited in the liver tissue induce egg granuloma formation and liver fibrosis, seriously threatening human health. Natural killer (NK) cells kill activated hepatic stellate cells (HSCs) or induce HSC apoptosis and inhibit the progression of liver fibrosis. However, the function of NK cells in liver fibrosis caused by S. japonicum infection is significantly inhibited. The mechanism of this inhibition remains unclear. Twenty mice were percutaneously infected with S. japonicum cercariae. Before infection and 2, 4, 6, and 8 weeks after infection, five mice were euthanized and dissected at each time point. Hepatic NK cells were isolated and transcriptome sequenced. The sequencing results showed that Tigit expression was high at 4-6 weeks post infection. This phenomenon was verified by reverse transcription quantitative PCR (RT-qPCR) and flow cytometry. NK cells derived from Tigit-/- and wild-type (WT) mice were co-cultured with HSCs. It was found that Tigit-/- NK cells induced apoptosis in a higher proportion of HSCs than WT NK cells. Schistosomiasis infection models of Tigit-/- and WT mice were established. The proportion and killing activity of hepatic NK cells were significantly higher in Tigit-/- mice than in WT mice. The degree of liver fibrosis in Tigit-/- mice was significantly lower than that in WT mice. NK cells were isolated from Tigit-/- and WT mice and injected via the tail vein into WT mice infected with S. japonicum. The degree of liver fibrosis in mice that received NK cell infusion reduced significantly, but there was no significant difference between mice that received NK cells from Tigit-/- and WT mice, respectively. Our findings indicate that Tigit knockout enhanced the function of NK cells and reduced the degree of liver fibrosis in schistosomiasis, thus providing a novel strategy for treating hepatic fibrosis induced by schistosomiasis.
Collapse
Affiliation(s)
- Yuan Gao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Xiaocheng Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Tingting Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Hao Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Hua Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Yuan Hu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
- * E-mail: (YH); (JC)
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (YH); (JC)
| |
Collapse
|
169
|
Wong DCP, Ding JL. The mechanobiology of NK cells- 'Forcing NK to Sense' target cells. Biochim Biophys Acta Rev Cancer 2023; 1878:188860. [PMID: 36791921 DOI: 10.1016/j.bbcan.2023.188860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 02/16/2023]
Abstract
Natural killer (NK) cells are innate immune lymphocytes that recognize and kill cancer and infected cells, which makes them unique 'off-the-shelf' candidates for a new generation of immunotherapies. Biomechanical forces in homeostasis and pathophysiology accrue additional immune regulation for NK immune responses. Indeed, cellular and tissue biomechanics impact NK receptor clustering, cytoskeleton remodeling, NK transmigration through endothelial cells, nuclear mechanics, and even NK-dendritic cell interaction, offering a plethora of unexplored yet important dynamic regulation for NK immunotherapy. Such events are made more complex by the heterogeneity of human NK cells. A significant question remains on whether and how biochemical and biomechanical cues collaborate for NK cell mechanotransduction, a process whereby mechanical force is sensed, transduced, and translated to downstream mechanical and biochemical signalling. Herein, we review recent advances in understanding how NK cells perceive and mechanotransduce biophysical cues. We focus on how the cellular cytoskeleton crosstalk regulates NK cell function while bearing in mind the heterogeneity of NK cells, the direct and indirect mechanical cues for NK anti-tumor activity, and finally, engineering advances that are of translational relevance to NK cell biology at the systems level.
Collapse
Affiliation(s)
- Darren Chen Pei Wong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore.
| | - Jeak Ling Ding
- Department of Biological Sciences, National University of Singapore, 117543, Singapore; Integrative Sciences and Engineering Programme, National University of Singapore, 119077, Singapore.
| |
Collapse
|
170
|
Ren J, Feng X, Guo Y, Kong D, Wang Y, Xiao J, Jiang W, Feng X, Liu X, Li A, Sun C, He M, Li B, Wang J, Jiang Y, Zheng C. GSK-3β/β-catenin pathway plays crucial roles in the regulation of NK cell cytotoxicity against myeloma cells. FASEB J 2023; 37:e22821. [PMID: 36794671 DOI: 10.1096/fj.202201658rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
The plasma cell malignancy, multiple myeloma (MM), has significantly improved by the application of new drugs and autologous hematopoietic stem cell transplantation. However, MM remains incurable. A number of studies have revealed an anti-MM effect of natural killer (NK) cells; however, their clinical efficacy is limited. Furthermore, glycogen synthase kinase (GSK)-3β inhibitors show an antitumor function. In this study, we aimed to evaluate the potential roles of a GSK-3β inhibitor (TWS119) in the regulation of NK cell cytotoxicity against MM. Our results showed that, in the presence of TWS119, the NK cell line, NK-92, and in vitro-expanded primary NK cells exhibited a significantly higher degranulation activity, expression of activating receptors, cellular cytotoxicity, and cytokine secretion when they were exposed to MM cells. Mechanistic studies indicated that TWS119 treatment markedly upregulated RAB27A expression, a key molecule for NK cell degranulation, and induced the colocalization of β-catenin with NF-κB in the nucleus of NK cells. More importantly, GSK-3β inhibition combined with the adoptive transfer of TWS119-treated NK-92 cells significantly reduced tumor volume and prolonged the survival time of myeloma-bearing mice. In summary, our novel findings suggest that targeting GSK-3β through the activation of β-catenin/NF-κB pathway may be an important approach to improve therapeutic efficacy of NK cell transfusion for MM.
Collapse
Affiliation(s)
- Jing Ren
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Xiumei Feng
- Department of Hematology, The Fourth People's Hospital of Jinan City, Jinan, Shandong, China
| | - Yanan Guo
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Dexiao Kong
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Yongjing Wang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Juan Xiao
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Wen Jiang
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoli Feng
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoli Liu
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Ai Li
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Congcong Sun
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Mingming He
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Bingen Li
- R&D Department, Weihai Zhengsheng Biotechnology Co., Ltd, Weihai, China
| | - Juandong Wang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Yang Jiang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China.,Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China
| |
Collapse
|
171
|
Korchagina AA, Koroleva E, Tumanov AV. Innate Lymphoid Cell Plasticity in Mucosal Infections. Microorganisms 2023; 11:461. [PMID: 36838426 PMCID: PMC9967737 DOI: 10.3390/microorganisms11020461] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Mucosal tissue homeostasis is a dynamic process that involves multiple mechanisms including regulation of innate lymphoid cells (ILCs). ILCs are mostly tissue-resident cells which are critical for tissue homeostasis and immune response against pathogens. ILCs can sense environmental changes and rapidly respond by producing effector cytokines to limit pathogen spread and initiate tissue recovery. However, dysregulation of ILCs can also lead to immunopathology. Accumulating evidence suggests that ILCs are dynamic population that can change their phenotype and functions under rapidly changing tissue microenvironment. However, the significance of ILC plasticity in response to pathogens remains poorly understood. Therefore, in this review, we discuss recent advances in understanding the mechanisms regulating ILC plasticity in response to intestinal, respiratory and genital tract pathogens. Key transcription factors and lineage-guiding cytokines regulate this plasticity. Additionally, we discuss the emerging data on the role of tissue microenvironment, gut microbiota, and hypoxia in ILC plasticity in response to mucosal pathogens. The identification of new pathways and molecular mechanisms that control functions and plasticity of ILCs could uncover more specific and effective therapeutic targets for infectious and autoimmune diseases where ILCs become dysregulated.
Collapse
Affiliation(s)
| | | | - Alexei V. Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA
| |
Collapse
|
172
|
Yang J, Tabuchi Y, Katsuki R, Taki M. bioTCIs: Middle-to-Macro Biomolecular Targeted Covalent Inhibitors Possessing Both Semi-Permanent Drug Action and Stringent Target Specificity as Potential Antibody Replacements. Int J Mol Sci 2023; 24:3525. [PMID: 36834935 PMCID: PMC9968108 DOI: 10.3390/ijms24043525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Monoclonal antibody therapies targeting immuno-modulatory targets such as checkpoint proteins, chemokines, and cytokines have made significant impact in several areas, including cancer, inflammatory disease, and infection. However, antibodies are complex biologics with well-known limitations, including high cost for development and production, immunogenicity, a limited shelf-life because of aggregation, denaturation, and fragmentation of the large protein. Drug modalities such as peptides and nucleic acid aptamers showing high-affinity and highly selective interaction with the target protein have been proposed alternatives to therapeutic antibodies. The fundamental limitation of short in vivo half-life has prevented the wide acceptance of these alternatives. Covalent drugs, also known as targeted covalent inhibitors (TCIs), form permanent bonds to target proteins and, in theory, eternally exert the drug action, circumventing the pharmacokinetic limitation of other antibody alternatives. The TCI drug platform, too, has been slow in gaining acceptance because of its potential prolonged side-effect from off-target covalent binding. To avoid the potential risks of irreversible adverse drug effects from off-target conjugation, the TCI modality is broadening from the conventional small molecules to larger biomolecules possessing desirable properties (e.g., hydrolysis resistance, drug-action reversal, unique pharmacokinetics, stringent target specificity, and inhibition of protein-protein interactions). Here, we review the historical development of the TCI made of bio-oligomers/polymers (i.e., peptide-, protein-, or nucleic-acid-type) obtained by rational design and combinatorial screening. The structural optimization of the reactive warheads and incorporation into the targeted biomolecules enabling a highly selective covalent interaction between the TCI and the target protein is discussed. Through this review, we hope to highlight the middle to macro-molecular TCI platform as a realistic replacement for the antibody.
Collapse
Affiliation(s)
- Jay Yang
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
- School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Department of GI Surgery II, Graduate School of Medicine, Hokkaido University, Sapporo 068-8638, Japan
| | - Yudai Tabuchi
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
| | - Riku Katsuki
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
| | - Masumi Taki
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
- Institute for Advanced Science, UEC, Chofu 182-8585, Japan
| |
Collapse
|
173
|
Kupke P, Adenugba A, Schemmerer M, Bitterer F, Schlitt HJ, Geissler EK, Wenzel JJ, Werner JM. Immunomodulation of Natural Killer Cell Function by Ribavirin Involves TYK-2 Activation and Subsequent Increased IFN-γ Secretion in the Context of In Vitro Hepatitis E Virus Infection. Cells 2023; 12:cells12030453. [PMID: 36766795 PMCID: PMC9913562 DOI: 10.3390/cells12030453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Hepatitis E virus (HEV) is a major cause of acute hepatitis globally. Chronic and fulminant courses are observed especially in immunocompromised transplant recipients since administration of ribavirin (RBV) does not always lead to a sustained virologic response. By in vitro stimulation of NK cells through hepatoma cell lines inoculated with a full-length HEV and treatment with RBV, we analyzed the viral replication and cell response to further elucidate the mechanism of action of RBV on immune cells, especially NK cells, in the context of HEV infection. Co-culture of HEV-infected hepatoma cells with PBMCs and treatment with RBV both resulted in a decrease in viral replication, which in combination showed an additive effect. An analysis of NK cell functions after stimulation revealed evidence of reduced cytotoxicity by decreased TRAIL and CD107a degranulation. Simultaneously, IFN-ɣ production was significantly increased through the IL-12R pathway. Although there was no direct effect on the IL-12R subunits, downstream events starting with TYK-2 and subsequently pSTAT4 were upregulated. In conclusion, we showed that RBV has an immunomodulatory effect on the IL-12R pathway of NK cells via TYK-2. This subsequently leads to an enhanced IFN-ɣ response and thus, to an additive antiviral effect in the context of an in vitro HEV infection.
Collapse
Affiliation(s)
- Paul Kupke
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Akinbami Adenugba
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Mathias Schemmerer
- National Consultant Laboratory for HAV and HEV, Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Florian Bitterer
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Hans J. Schlitt
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Edward K. Geissler
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Jürgen J. Wenzel
- National Consultant Laboratory for HAV and HEV, Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Jens M. Werner
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
- Correspondence:
| |
Collapse
|
174
|
Singh L, Nair L, Kumar D, Arora MK, Bajaj S, Gadewar M, Mishra SS, Rath SK, Dubey AK, Kaithwas G, Choudhary M, Singh M. Hypoxia induced lactate acidosis modulates tumor microenvironment and lipid reprogramming to sustain the cancer cell survival. Front Oncol 2023; 13:1034205. [PMID: 36761981 PMCID: PMC9906992 DOI: 10.3389/fonc.2023.1034205] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023] Open
Abstract
It is well known that solid hypoxic tumour cells oxidise glucose through glycolysis, and the end product of this pathway is fermented into lactate which accumulates in the tumour microenvironment (TME). Initially, it was proclaimed that cancer cells cannot use lactate; therefore, they dump it into the TME and subsequently augment the acidity of the tumour milieu. Furthermore, the TME acts as a lactate sink with stope variable amount of lactate in different pathophysiological condition. Regardless of the amount of lactate pumped out within TME, it disappears immediately which still remains an unresolved puzzle. Recent findings have paved pathway in exploring the main role of lactate acidosis in TME. Cancer cells utilise lactate in the de novo fatty acid synthesis pathway to initiate angiogenesis and invasiveness, and lactate also plays a crucial role in the suppression of immunity. Furthermore, lactate re-programme the lipid biosynthetic pathway to develop a metabolic symbiosis in normoxic, moderately hypoxic and severely hypoxic cancer cells. For instance: severely hypoxic cancer cells enable to synthesizing poly unsaturated fatty acids (PUFA) in oxygen scarcity secretes excess of lactate in TME. Lactate from TME is taken up by the normoxic cancer cells whereas it is converted back to PUFAs after a sequence of reactions and then liberated in the TME to be utilized in the severely hypoxic cancer cells. Although much is known about the role of lactate in these biological processes, the exact molecular pathways that are involved remain unclear. This review attempts to understand the molecular pathways exploited by lactate to initiate angiogenesis, invasiveness, suppression of immunity and cause re-programming of lipid synthesis. This review will help the researchers to develop proper understanding of lactate associated bimodal regulations of TME.
Collapse
Affiliation(s)
- Lakhveer Singh
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Lakshmi Nair
- Department of Pharmaceutical Science, Assam University (A Central University), Silchar, Assam, India
| | - Dinesh Kumar
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India
| | - Mandeep Kumar Arora
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Sakshi Bajaj
- Chaudhary Devi Lal College of Pharmacy, Yamuna Nagar, India
| | - Manoj Gadewar
- School of Medical and Allied Sciences, KR Mangalam University, Gurgaon, India
| | | | - Santosh Kumar Rath
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Amit Kumar Dubey
- School of Pharmaceutical & Population Health Informatics, DIT University, Dehradun, India
| | - Gaurav Kaithwas
- Department of Pharmaceutical Science, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Manjusha Choudhary
- University Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, India,*Correspondence: Manjusha Choudhary, ; Manjari Singh,
| | - Manjari Singh
- Department of Pharmaceutical Science, Assam University (A Central University), Silchar, Assam, India,*Correspondence: Manjusha Choudhary, ; Manjari Singh,
| |
Collapse
|
175
|
Zhu Y, Shi J. Cytotoxic and chemotactic dynamics of NK cells quantified by live-cell imaging. Methods Cell Biol 2023; 173:49-64. [PMID: 36653085 DOI: 10.1016/bs.mcb.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Natural Killer (NK) cells detect and eliminate virus-infected cells and cancer cells, and are crucial players of the human immune defense system. Although the relevant molecular machineries involved in NK cell activation and NK-target cell interactions are largely known, how their collective signaling modulates the dynamic behaviors of NK cells, e.g., motility and cytotoxicity, and the rate-limiting kinetics involved are still in need of comprehensive investigations. In traditional bulk killing assays, heterogeneity and kinetic details of individual NK-target cell interactions are masked, seriously limiting analysis of the underlying dynamic mechanisms. Here we present detailed protocols of a number of live-cell imaging assays using fluorescent protein reporters and/or a live-cell dye that enable the acquisition of quantitative kinetic data at the single cell level for elucidating the mechanism underlying the interaction dynamics of primary human NK cells and epithelial cancer cells. Moreover, we discuss how the imaging data can be analyzed either alone or in combination to quantify and determine the key dynamic steps/intermediates involved in specific NK cell activity, e.g., NK cell cytotoxic modes and their associated kinetics, and NK cell motility toward different cancer targets. These live-cell imaging assays can be easily adapted to analyze the rate-limiting kinetics and heterogeneity of other cell-cell interaction dynamics, e.g., in T cell function.
Collapse
Affiliation(s)
- Yanting Zhu
- Department of Physics and Department of Biology, Center for Quantitative Systems Biology, Hong Kong Baptist University, Hong Kong, China
| | - Jue Shi
- Department of Physics and Department of Biology, Center for Quantitative Systems Biology, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
176
|
Terrén I, Astarloa-Pando G, Amarilla-Irusta A, Borrego F. P815-based redirected degranulation assay to study human NK cell effector functions. Methods Cell Biol 2023; 173:33-48. [PMID: 36653084 DOI: 10.1016/bs.mcb.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Natural killer (NK) cells are part of the innate immune system, the classic cytotoxic population of innate lymphoid cells (ILCs). They can directly kill virus-infected or tumor cells through different mechanisms without prior sensitization using their lytic functions in response to different signals (target cell ligands and/or inflammatory cytokines) and secreting cytokines, such as interferon gamma (IFNγ) and tumor necrosis factor (TNF). NK cells use antibody-dependent cell-mediated cytotoxicity (ADCC) to recognize and kill cells expressing target antigens when they are antibody coated. Redirected cytotoxicity is a technique used to target cells that do not per se activate NK cells. Here, we use redirected degranulation, a surrogate technique that correlates with redirected lysis. The P815 cell line (mouse mastocytoma) express fragment crystallizable gamma receptor II (FcγRII) and therefore could bind the Fc portion of mouse IgG antibodies, which through their fragment antigen-binding (Fab) may recognize NK cells activating receptors leading to target cell lysis. This technique could be used to determine the inhibitory or activating capacity of different receptors or isoforms and in immunotherapy using T cell and NK cell activators.
Collapse
Affiliation(s)
- Iñigo Terrén
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, Barakaldo, Spain
| | | | | | - Francisco Borrego
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
177
|
Ye Z, Zhang H, Liang J, Yi S, Zhan X. Significance of logistic regression scoring model based on natural killer cell-mediated cytotoxic pathway in the diagnosis of colon cancer. Front Immunol 2023; 14:1117908. [PMID: 36742322 PMCID: PMC9895796 DOI: 10.3389/fimmu.2023.1117908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
Background The poor clinical accuracy to predict the survival of colon cancer patients is associated with a high incidence rate and a poor 3-year survival rate. This study aimed to identify the poor prognostic biomarkers of colon cancer from natural killer cell-mediated cytotoxic pathway (NKCP), and establish a logistical regression scoring model to predict its prognosis. Methods Based on the expressions and methylations of NKCP-related genes (NRGs) and the clinical information, dimensionality reduction screening was performed to establish a logistic regression scoring model to predict survival and prognosis. Risk score, clinical stage, and ULBP2 were used to establish a logistic regression scoring model to classify the 3-year survival period and compare with each other. Comparison of survival, tumor mutation burden (TMB), estimation of immune invasion, and prediction of chemotherapeutic drug IC50 were performed between low- and high-risk score groups. Results This study found that ULBP2 was significantly overexpressed in colon cancer tissues and colon cancer cell lines. The logistic regression scoring model was established to include six statistically significant features: S = 1.70 × stage - 9.32 × cg06543087 + 6.19 × cg25848557 + 1.29 × IFNA1 + 0.048 × age + 4.37 × cg21370856 - 8.93, which was used to calculate risk score of each sample. The risk scores, clinical stage, and ULBP2 were classified into three-year survival, the 3-year prediction accuracy based on 10-fold cross-validation was 80.17%, 67.24, and 59.48%, respectively. The survival time of low-risk score group was better than that of the high-risk score group. Moreover, compared to high-risk score group, low-risk score group had lower TMB [2.20/MB (log10) vs. 2.34/MB (log10)], higher infiltration score of M0 macrophages (0.17 vs. 0.14), and lower mean IC50 value of oxaliplatin (3.65 vs 3.78) (p < 0.05). Conclusions The significantly upregulated ULBP2 was a poor prognostic biomarker of colon cancer. The risk score based on the six-feature logistic regression model can effectively predict the 3-year survival time. High-risk score group demonstrated a poorer prognosis, higher TMB, lower M0 macrophage infiltration score, and higher IC50 value of oxaliplatin. The six-feature logistic scoring model has certain clinical significance in colon cancer.
Collapse
Affiliation(s)
- Zhen Ye
- Medical Science and Technology Innovation Center, The Second Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Huanhuan Zhang
- Medical Science and Technology Innovation Center, The Second Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Jianwei Liang
- Department of General Surgery, Tai ‘an Central Hospital, Taian, Shandong, China
| | - Shuying Yi
- Medical Science and Technology Innovation Center, The Second Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China,*Correspondence: Xianquan Zhan, ; Shuying Yi,
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, The Second Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China,*Correspondence: Xianquan Zhan, ; Shuying Yi,
| |
Collapse
|
178
|
Pathogenesis of Anemia in Canine Babesiosis: Possible Contribution of Pro-Inflammatory Cytokines and Chemokines-A Review. Pathogens 2023; 12:pathogens12020166. [PMID: 36839438 PMCID: PMC9962459 DOI: 10.3390/pathogens12020166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Canine babesiosis is a tick-borne protozoan disease caused by intraerythrocytic parasites of the genus Babesia. The infection may lead to anemia in infected dogs. However, anemia is not directly caused by the pathogen. The parasite's developmental stages only have a marginal role in contributing to a decreased red blood cell (RBC) count. The main cause of anemia in affected dogs is the immune response to the infection. This response includes antibody production, erythrophagocytosis, oxidative damage of RBCs, complement activation, and antibody-dependent cellular cytotoxicity. Moreover, both infected and uninfected erythrocytes are retained in the spleen and sequestered in micro-vessels. All these actions are driven by pro-inflammatory cytokines and chemokines, especially IFN-γ, TNF-α, IL-6, and IL-8. Additionally, imbalance between the actions of pro- and anti-inflammatory cytokines plays a role in patho-mechanisms leading to anemia in canine babesiosis. This article is a review of the studies on the pathogenesis of anemia in canine babesiosis and related diseases, such as bovine or murine babesiosis and human or murine malaria, and the role of pro-inflammatory cytokines and chemokines in the mechanisms leading to anemia in infected dogs.
Collapse
|
179
|
Lu C, Liu Y, Ali NM, Zhang B, Cui X. The role of innate immune cells in the tumor microenvironment and research progress in anti-tumor therapy. Front Immunol 2023; 13:1039260. [PMID: 36741415 PMCID: PMC9893925 DOI: 10.3389/fimmu.2022.1039260] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/14/2022] [Indexed: 01/20/2023] Open
Abstract
Innate immune cells in the tumor microenvironment (TME) mainly include macrophages, neutrophils, natural killer cells, dendritic cells and bone marrow derived suppressor cells. They play an anti-tumor or pro-tumor role by secreting various cytokines, chemokines and other factors, and determine the occurrence and development of tumors. Comprehending the role of innate immune cells in tumorigenesis and progression can help improve therapeutic approaches targeting innate immune cells in the TME, increasing the likelihood of favorable prognosis. In this review, we discussed the cell biology of innate immune cells, their role in tumorigenesis and development, and the current status of innate immune cell-based immunotherapy, in order to provide an overview for future research lines and clinical trials.
Collapse
Affiliation(s)
- Chenglin Lu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China,Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Nasra Mohamoud Ali
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China,*Correspondence: Xiaonan Cui, ; Bin Zhang,
| | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China,*Correspondence: Xiaonan Cui, ; Bin Zhang,
| |
Collapse
|
180
|
Mechanisms of Neuroinvasion and Neuropathogenesis by Pathologic Flaviviruses. Viruses 2023; 15:v15020261. [PMID: 36851477 PMCID: PMC9965671 DOI: 10.3390/v15020261] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/07/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Flaviviruses are present on every continent and cause significant morbidity and mortality. In many instances, severe cases of infection with flaviviruses involve the invasion of and damage to the central nervous system (CNS). Currently, there are several mechanisms by which it has been hypothesized flaviviruses reach the brain, including the disruption of the blood-brain barrier (BBB) which acts as a first line of defense by blocking the entry of many pathogens into the brain, passing through the BBB without disruption, as well as travelling into the CNS through axonal transport from peripheral nerves. After flaviviruses have entered the CNS, they cause different neurological symptoms, leading to years of neurological sequelae or even death. Similar to neuroinvasion, there are several identified mechanisms of neuropathology, including direct cell lysis, blockage of the cell cycle, indication of apoptosis, as well as immune induced pathologies. In this review, we aim to summarize the current knowledge in the field of mechanisms of both neuroinvasion and neuropathogenesis during infection with a variety of flaviviruses and examine the potential contributions and timing of each discussed pathway.
Collapse
|
181
|
Akrami S, Tahmasebi A, Moghadam A, Ramezani A, Niazi A. Integration of mRNA and protein expression data for the identification of potential biomarkers associated with pancreatic ductal adenocarcinoma. Comput Biol Med 2023; 157:106529. [PMID: 36921457 DOI: 10.1016/j.compbiomed.2022.106529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/21/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most death-dealing tumors, with a tremendously poor prognosis. Here, we, through interrogation of mRNA and protein data combined with a system biology approach, identified several key genes, functional processes, and pathways that can have critical roles in PDAC. We detected an interesting module related to the clinical traits that enriched in the ribosome, hematopoietic cell lineage, and cell adhesion molecules-related pathways. We also identified several hub genes in important modules that are associated with immune system processes. The results also indicated some lncRNAs, such as FAM30A, and MIR223HG with essential functions that are involved in PDAC. Additionally, five genes, including CD53, ITGAL, WDFY4, TLX1, and LMAN1L were screened by survival analysis and can be considered as candidate biomarkers or therapeutic targets. According to our strategy, the findings of this study may provide a better understanding of the molecular mechanisms and suggest potential prognostic and therapeutic targets for PDAC.
Collapse
Affiliation(s)
- Sahar Akrami
- Institute of Biotechnology, Shiraz University, Shiraz, Iran
| | - Ahmad Tahmasebi
- Institute of Biotechnology, Shiraz University, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Moghadam
- Institute of Biotechnology, Shiraz University, Shiraz, Iran
| | - Amin Ramezani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Niazi
- Institute of Biotechnology, Shiraz University, Shiraz, Iran.
| |
Collapse
|
182
|
He XQ, Qiu HQ, Wang M, Mao YF, Li XY, Wang XY, Geng YL, Wang L. Uncorrected Preoperative Infection Causing the Death of a Patient with a Thoracic Aortic Aneurysm. Infect Drug Resist 2023; 16:243-248. [PMID: 36660348 PMCID: PMC9842481 DOI: 10.2147/idr.s396269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023] Open
Abstract
Background A thoracic aortic aneurysm (TAA) is a known condition seen in cardiovascular practice. A TAA rupture and postoperative infection may result in death. Preoperative infections leading to death are extremely rare. Case Study A 62-year-old Chinese female was admitted to The First Hospital of Hebei Medical University with a two-day history of abdominal pain. She was diagnosed with a TAA rupture and underwent immediate surgery. The preoperative urine analysis indicated that the positive bacteria and white blood cell count suggested a urinary tract bacterial infection. The patient was administered the empiric antibiotics, cefazolin; however, her blood pressure continued to drop during the perioperative period and she died of uncorrectable acidosis 8 h after the operation. On the second day after death, both the blood and urine cultures were positive for Pseudomonas aeruginosa. Conclusion Given that this patient with a TAA rupture died of uncorrected acidosis caused by preoperative infection, it is important to evoke the diagnosis in the context of TAA. Routine laboratory indicators are valuable factors for surgeons and physicians in assessing a patient's condition and improving their prognosis.
Collapse
Affiliation(s)
- Xin-Qi He
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Hui-Qing Qiu
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Meng Wang
- Department of Radiology and Nuclear Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Ya-Fei Mao
- Department of Laboratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xin-Yuan Li
- Department of Laboratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xian-Yun Wang
- Scientific Research Data Center, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Yu-Lan Geng
- Department of Laboratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China,Correspondence: Yu-Lan Geng, Department of Laboratory Medicine, The First Hospital of Hebei Medical University, No. 89 of Donggang Road, Yuhua District, Shijiazhuang, 050031, People’s Republic of China, Tel +86 311 87156567, Fax +86 311 85917029, Email
| | - Le Wang
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China,Le Wang, Department of Cardiology, The First Hospital of Hebei Medical University, No. 89 of Donggang Road, Yuhua District, Shijiazhuang, 050031, People’s Republic of China, Tel +86 311 87155263, Fax +86 311 85917029, Email
| |
Collapse
|
183
|
Targeting Epigenetic Mechanisms: A Boon for Cancer Immunotherapy. Biomedicines 2023; 11:biomedicines11010169. [PMID: 36672677 PMCID: PMC9855697 DOI: 10.3390/biomedicines11010169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Immunotherapy is rapidly emerging as a promising approach against cancer. In the last decade, various immunological mechanisms have been targeted to induce an increase in the immune response against cancer cells. However, despite promising results, many patients show partial response, resistance, or serious toxicities. A promising way to overcome this is the use of immunotherapeutic approaches, in combination with other potential therapeutic approaches. Aberrant epigenetic modifications play an important role in carcinogenesis and its progression, as well as in the functioning of immune cells. Thus, therapeutic approaches targeting aberrant epigenetic mechanisms and the immune response might provide an effective antitumor effect. Further, the recent development of potent epigenetic drugs and immunomodulators gives hope to this combinatorial approach. In this review, we summarize the synergy mechanism between epigenetic therapies and immunotherapy for the treatment of cancer, and discuss recent advancements in the translation of this approach.
Collapse
|
184
|
Wang ZH, Li W, Dong H, Han F. Current state of NK cell-mediated immunotherapy in chronic lymphocytic leukemia. Front Oncol 2023; 12:1077436. [PMID: 37078002 PMCID: PMC10107371 DOI: 10.3389/fonc.2022.1077436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) has become one of the most common hematological diseases in western countries, with an annual incidence of 42/100,000. Conventional chemotherapy and targeted therapeutic drugs showed limitations in prognosis or in efficiency in high-risk patients. Immunotherapy represented is one of the most effective therapeutic approaches with the potential of better effect and prognosis. Natural killer (NK) cells are good options for immunotherapy as they can effectively mediate anti-tumor activity of immune system by expressing activating and inhibiting receptors and recognizing specific ligands on various tumor cells. NK cells are critical in the immunotherapy of CLL by enhancing self-mediated antibody-dependent cytotoxicity (ADCC), allogeneic NK cell therapy and chimeric antigen receptor-natural killer (CAR-NK) cell therapy. In this article, we reviewed the features, working mechanisms, and receptors of NK cells, and the available evidence of the advantages and disadvantages of NK cell-based immunotherapies, and put forward future study directions in this field.
Collapse
Affiliation(s)
- Zong-Han Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Li
- Department of General Surgery, Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Hao Dong
- Department of Gastrointestinal Nutrition and Surgical Surgery, The Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Hao Dong, ; Fujun Han,
| | - Fujun Han
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Hao Dong, ; Fujun Han,
| |
Collapse
|
185
|
Wang Q, Li X, Qiu J, He Y, Wu J, Li J, Liu W, Han J. A pathway-based mutation signature to predict the clinical outcomes and response to CTLA-4 inhibitors in melanoma. Comput Struct Biotechnol J 2023; 21:2536-2546. [PMID: 37102155 PMCID: PMC10123336 DOI: 10.1016/j.csbj.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/28/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has become a powerful clinical strategy for treating melanoma. The relationship between somatic mutations and the clinical benefits of immunotherapy has been widely recognized. However, the gene-based predictive biomarkers are less stable due to the heterogeneity of cancer at the individual gene level. Recent studies have suggested that the accumulation of gene mutations in biological pathways may activate antitumor immune responses. Herein, a novel pathway mutation signature (PMS) was constructed to predict the survival and efficacy of ICI therapy. In a dataset of melanoma patients treated with anti-CTLA-4, we mapped the mutated genes into the pathways and then identified seven significant mutation pathways associated with survival and immunotherapy response, which were used to construct the PMS model. According to the PMS model, the patients in the PMS-high group showed better overall survival (hazard ratio (HR) = 0.37; log-rank test, p < 0.0001) and progression-free survival (HR = 0.52; log-rank test, p = 0.014) than those in the PMS-low group. The PMS-high patients also showed a significantly higher objective response rate to anti-CTLA-4 therapy than the PMS-low patients (Fisher's exact test, p = 0.0055), and the predictive power of the PMS model was superior to that of TMB. Finally, the prognostic and predictive value of the PMS model was validated in two independent validation sets. Our study demonstrated that the PMS model can be considered a potential biomarker to predict the clinical outcomes and response to anti-CTLA-4 therapy in melanoma patients.
Collapse
Affiliation(s)
- Qian Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Xiangmei Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Jiayue Qiu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Yalan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Jiashuo Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Ji Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Wei Liu
- College of Science, Heilongjiang Institute of Technology, Harbin 150050, PR China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| |
Collapse
|
186
|
Alicata C, Veneziani I, Ricci B, Moretta L, Maggi E. Determining Endosomal Toll-Like Receptors Gene Expression in NK Cells After Stimulation with Specific Agonists. Methods Mol Biol 2023; 2700:151-162. [PMID: 37603179 DOI: 10.1007/978-1-0716-3366-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Poor knowledge is currently available about the biology of Toll-like receptors (TLRs) in natural killer (NK) cells. This is particularly due to the old belief that NK cells are unable to specifically eliminate microbes without presensitization. On the contrary, it has been clearly demonstrated that not only they can be activated through the engagement of Toll-like receptors (TLRs) by microbial molecules, but also that this interaction induces NK cells to release cytokines that, in turn, activate other cells of both innate and adaptive immunity. For this reason, immunotherapy based on local infusion of TLRs ligands is currently considered as a novel potential strategy to treat solid tumors. Here, we provide a protocol to efficiently stimulate NK cells via endosomal TLRs agonists and to determine endosomal TLRs gene expression level. This protocol can be used for in vitro investigation into endosomal TLRs function in NK cells under different conditions.
Collapse
Affiliation(s)
- Claudia Alicata
- Tumor Immunology Unit, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy.
| | - Irene Veneziani
- Tumor Immunology Unit, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Biancamaria Ricci
- Tumor Immunology Unit, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Enrico Maggi
- Tumor Immunology Unit, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| |
Collapse
|
187
|
Calvo-Schimmel A, Kober KM, Paul SM, Cooper BA, Harris C, Shin J, Hammer MJ, Conley YP, Dokiparthi V, Olshen A, Levine JD, Miaskowski C. Sleep disturbance is associated with perturbations in immune-inflammatory pathways in oncology outpatients undergoing chemotherapy. Sleep Med 2023; 101:305-315. [PMID: 36470166 PMCID: PMC11200329 DOI: 10.1016/j.sleep.2022.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE/BACKGROUND Sleep disturbance is a common problem in patients receiving chemotherapy. Purpose was to evaluate for perturbations in immune-inflammatory pathways between oncology patients with low versus very high levels of sleep disturbance. PATIENTS/METHODS Sleep disturbance was evaluated using the General Sleep Disturbance Scale six times over two cycles of chemotherapy. Latent profile analysis was used to identify subgroups of patients with distinct sleep disturbance profiles. Pathway impact analyses were performed in two independent samples using gene expression data obtained from RNA sequencing (n = 198) and microarray (n = 162) technologies. Fisher's combined probability test was used to identify significantly perturbed pathways between Low versus Very High sleep disturbance classes. RESULTS In the RNA sequencing and microarray samples, 59.1% and 51.9% of patients were in the Very High sleep disturbance class, respectively. Thirteen perturbed pathways were related to immune-inflammatory mechanisms (i.e., endocytosis, phagosome, antigen processing and presentation, natural killer cell mediated cytotoxicity, cytokine-cytokine receptor interaction, apoptosis, neutrophil extracellular trap formation, nucleotide-binding and oligomerization domain-like receptor signaling, Th17 cell differentiation, intestinal immune network for immunoglobulin A production, T-cell receptor signaling, complement and coagulation cascades, and tumor necrosis factor signaling). CONCLUSIONS First study to identify perturbations in immune-inflammatory pathways associated with very high levels of sleep disturbance in oncology outpatients. Findings suggest that complex immune-inflammatory interactions underlie sleep disturbance.
Collapse
Affiliation(s)
- Alejandra Calvo-Schimmel
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Kord M Kober
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Steven M Paul
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Bruce A Cooper
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Carolyn Harris
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Joosun Shin
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | | | - Yvette P Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Vasuda Dokiparthi
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| | - Adam Olshen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.
| | - Jon D Levine
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Christine Miaskowski
- Department of Physiological Nursing, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
188
|
Wang N, Zhao L, Zhang D, Kong F. Research progress on the immunomodulatory mechanism of acupuncture in tumor immune microenvironment. Front Immunol 2023; 14:1092402. [PMID: 36865562 PMCID: PMC9971227 DOI: 10.3389/fimmu.2023.1092402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
With the constantly deeper understanding of individualized precision therapy, immunotherapy is increasingly developed and personalized. The tumor immune microenvironment (TIME) mainly consists of infiltrating immune cells, neuroendocrine cells, extracellular matrix, lymphatic vessel network, etc. It is the internal environment basis for the survival and development of tumor cells. As a characteristic treatment of traditional Chinese medicine, acupuncture has shown potentially beneficial impacts on TIME. The currently available information demonstrated that acupuncture could regulate the state of immunosuppression through a range of pathways. An effective way to understand the mechanisms of action of acupuncture was to analyze the response following treatment of the immune system. This research reviewed the mechanisms of acupuncture regulating tumor immunological status based on innate and adaptive immunity.
Collapse
Affiliation(s)
- Na Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lu Zhao
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dou Zhang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Fanming Kong
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
189
|
Zhang Y, Zhao Y, Zhai Y, He J, Tang M, Liu Y, Yao Y, Xue P, He M, Li Q, Xu Y, Qu W, Zhang Y. Cadmium impairs the development of natural killer cells and bidirectionally modifies their capacity for cytotoxicity. CHEMOSPHERE 2023; 311:137068. [PMID: 36330983 DOI: 10.1016/j.chemosphere.2022.137068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
Cadmium (Cd) is a highly toxic heavy metal in the environment. The aim of this study was to investigate the impact of Cd on natural killer (NK) cells. C57BL/6 mice were treated with 10 ppm Cd via drinking water for 3 months, and the development of NK cells in the bone marrow (BM) and the cytotoxicity of mature NK (mNK) cells in the peripheral immune organs were evaluated thereafter; the impact of Cd on the cytotoxicity of mNK cells from human peripheral blood mononuclear cells (PBMC) was also investigated. Whereas Cd treatment impaired the differentiation of NK progenitors in the BM, Cd treatment activated the JAK3/STAT5 signaling to drive the proliferation of mNK cells and thereby lead to a compensation increase of mNK cells in the peripheral immune organs of mice. Additionally, Cd treatment bidirectionally regulated the cytotoxicity of mouse mNK cells to differential tumor cells, dependent on the levels of Fas expression in the tumor cells; mechanically, Cd treatment activated the JAK3/STAT5 signaling to promote the expression of FasL in mNK cells to increase their cytotoxicity, while Cd treatment reduced the expression of granzyme B in mNK cells to impair their cytotoxicity in the peripheral immune organs of mice. Likewise, in vitro assays indicated that Cd treatment also activated the JAK3/STAT5 signaling to increase the expression of FasL, whereas Cd treatment reduced the expression of granzyme B in human mNK cells. Thus Cd treatment impaired the development of NK cells in the BM and bidirectionally regulated the cytotoxicity of mNK cells in the peripheral immune organs, which may extend our current understanding for the immunotoxicity of Cd.
Collapse
Affiliation(s)
- Yufan Zhang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Yifan Zhao
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Yue Zhai
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Jinyi He
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Mengke Tang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Yalin Liu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Ye Yao
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Peng Xue
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Qian Li
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yanyi Xu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Weidong Qu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Yubin Zhang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
190
|
Whalen KA, Rakhra K, Mehta NK, Steinle A, Michaelson JS, Baeuerle PA. Engaging natural killer cells for cancer therapy via NKG2D, CD16A and other receptors. MAbs 2023; 15:2208697. [PMID: 37165468 PMCID: PMC10173799 DOI: 10.1080/19420862.2023.2208697] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023] Open
Abstract
The field of immuno-oncology has revolutionized cancer patient care and improved survival and quality of life for patients. Much of the focus in the field has been on exploiting the power of the adaptive immune response through therapeutic targeting of T cells. While these approaches have markedly advanced the field, some challenges remain, and the clinical benefit of T cell therapies does not extend to all patients or tumor indications. Alternative strategies, such as engaging the innate immune system, have become an intense area of focus in the field. In particular, the engagement of natural killer (NK) cells as potent effectors of the innate immune response has emerged as a promising modality in immunotherapy. Here, we review therapeutic approaches for selective engagement of NK cells for cancer therapy, with a particular focus on targeting the key activating receptors NK Group 2D (NKG2D) and cluster of differentiation 16A (CD16A).
Collapse
Affiliation(s)
- Kerry A. Whalen
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Kavya Rakhra
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Naveen K. Mehta
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
- Preclinical and Early Development, Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | | | - Patrick A. Baeuerle
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
- Institute for Immunology, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
191
|
Wang F, Cui Y, He D, Gong L, Liang H. Natural killer cells in sepsis: Friends or foes? Front Immunol 2023; 14:1101918. [PMID: 36776839 PMCID: PMC9909201 DOI: 10.3389/fimmu.2023.1101918] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Sepsis is one of the major causes of death in the hospital worldwide. The pathology of sepsis is tightly associated with dysregulation of innate immune responses. The contribution of macrophages, neutrophils, and dendritic cells to sepsis is well documented, whereas the role of natural killer (NK) cells, which are critical innate lymphoid lineage cells, remains unclear. In some studies, the activation of NK cells has been reported as a risk factor leading to severe organ damage or death. In sharp contrast, some other studies revealed that triggering NK cell activity contributes to alleviating sepsis. In all, although there are several reports on NK cells in sepsis, whether they exert detrimental or protective effects remains unclear. Here, we will review the available experimental and clinical studies about the opposing roles of NK cells in sepsis, and we will discuss the prospects for NK cell-based immunotherapeutic strategies for sepsis.
Collapse
Affiliation(s)
- Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yiqin Cui
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dongmei He
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lisha Gong
- School of Laboratory Medicine and Technology, Harbin Medical University, Daqing, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
192
|
Polten R, Kutle I, Hachenberg J, Klapdor R, Morgan M, Schambach A. Towards Novel Gene and Cell Therapy Approaches for Cervical Cancer. Cancers (Basel) 2022; 15:cancers15010263. [PMID: 36612258 PMCID: PMC9818159 DOI: 10.3390/cancers15010263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Cervical cancer is one of the most common malignancies in women, and the majority of cases are caused by infection with high-risk human papilloma virus (HPV) subtypes. Despite effective preventative measures, such as vaccinations against HPV, over 300,000 women die world-wide from cervical cancer each year. Once cervical cancer is diagnosed, treatment may consist of radial hysterectomy, or chemotherapy and radiotherapy, or a combination of therapies dependent upon the disease stage. Unfortunately, overall prognosis for patients with metastatic or recurrent disease remains poor. In these cases, immunotherapies may be useful based on promising preclinical work, some of which has been successfully translated to the clinic. For example, approaches using monoclonal antibodies directed against surface proteins important for control of immune checkpoints (i.e., immune checkpoint inhibitors) were shown to improve outcome in many cancer settings, including cervical cancer. Additionally, initial clinical studies showed that application of cytotoxic immune cells modified to express chimeric antigen receptors (CAR) or T cell receptors (TCR) for better recognition and elimination of tumor cells may be useful to control cervical cancer. This review explores these important topics, including strengths and limitations of standard and developing approaches, and how some novel treatment strategies may be optimally used to offer the best possible treatment for cervical cancer patients.
Collapse
Affiliation(s)
- Robert Polten
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Ivana Kutle
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Hachenberg
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| | - Rüdiger Klapdor
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence: (M.M.); (A.S.); Tel.: +49-511-532-6067 (A.S.)
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (M.M.); (A.S.); Tel.: +49-511-532-6067 (A.S.)
| |
Collapse
|
193
|
CAR-NK as a Rapidly Developed and Efficient Immunotherapeutic Strategy against Cancer. Cancers (Basel) 2022; 15:cancers15010117. [PMID: 36612114 PMCID: PMC9817948 DOI: 10.3390/cancers15010117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T cell therapy has been rapidly developing in recent years, ultimately revolutionizing immunotherapeutic strategies and providing significant anti-tumor potency, mainly in treating hematological neoplasms. However, graft-versus-host disease (GVHD) and other adverse effects, such as cytokine release syndromes (CRS) and neurotoxicity associated with CAR-T cell infusion, have raised some concerns about the broad application of this therapy. Natural killer (NK) cells have been identified as promising alternative platforms for CAR-based therapies because of their unique features, such as a lack of human leukocyte antigen (HLA)-matching restriction, superior safety, and better anti-tumor activity when compared with CAR-T cells. The lack of CRS, neurotoxicity, or GVHD, in the case of CAR-NK therapy, in addition to the possibility of using allogeneic NK cells as a CAR platform for "off-the-shelf" therapy, opens new windows for strategic opportunities. This review underlines recent design achievements in CAR constructs and summarizes preclinical studies' results regarding CAR-NK therapies' safety and anti-tumor potency. Additionally, new approaches in CAR-NK technology are briefly described, and currently registered clinical trials are listed.
Collapse
|
194
|
Raja R, Wu C, Bassoy EY, Rubino TE, Utagawa EC, Magtibay PM, Butler KA, Curtis M. PP4 inhibition sensitizes ovarian cancer to NK cell-mediated cytotoxicity via STAT1 activation and inflammatory signaling. J Immunother Cancer 2022; 10:jitc-2022-005026. [PMID: 36564125 PMCID: PMC9791393 DOI: 10.1136/jitc-2022-005026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Increased infiltration of T cells into ovarian tumors has been repeatedly shown to be predictive of enhanced patient survival. However, despite the evidence of an active immune response in ovarian cancer (OC), the frequency of responses to immune checkpoint blockade (ICB) therapy in OC is much lower than other cancer types. Recent studies have highlighted that deficiencies in the DNA damage response (DDR) can drive increased genomic instability and tumor immunogenicity, which leads to enhanced responses to ICB. Protein phosphatase 4 (PP4) is a critical regulator of the DDR; however, its potential role in antitumor immunity is currently unknown. RESULTS Our results show that the PP4 inhibitor, fostriecin, combined with carboplatin leads to increased carboplatin sensitivity, DNA damage, and micronuclei formation. Using multiple OC cell lines, we show that PP4 inhibition or PPP4C knockdown combined with carboplatin triggers inflammatory signaling via Nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 1 (STAT1) activation. This resulted in increased expression of the pro-inflammatory cytokines and chemokines: CCL5, CXCL10, and IL-6. In addition, IFNB1 expression was increased suggesting activation of the type I interferon response. Conditioned media from OC cells treated with the combination of PP4 inhibitor and carboplatin significantly increased migration of both CD8 T cell and natural killer (NK) cells over carboplatin treatment alone. Knockdown of stimulator of interferon genes (STING) in OC cells significantly abrogated the increase in CD8 T-cell migration induced by PP4 inhibition. Co-culture of NK-92 cells and OC cells with PPP4C or PPP4R3B knockdown resulted in strong induction of NK cell interferon-γ, increased degranulation, and increased NK cell-mediated cytotoxicity against OC cells. Stable knockdown of PP4C in a syngeneic, immunocompetent mouse model of OC resulted in significantly reduced tumor growth in vivo. Tumors with PP4C knockdown had increased infiltration of NK cells, NK T cells, and CD4+ T cells. Addition of low dose carboplatin treatment led to increased CD8+ T-cell infiltration in PP4C knockdown tumors as compared with the untreated PP4C knockdown tumors. CONCLUSIONS Our work has identified a role for PP4 inhibition in promoting inflammatory signaling and enhanced immune cell effector function. These findings support the further investigation of PP4 inhibitors to enhance chemo-immunotherapy for OC treatment.
Collapse
Affiliation(s)
- Remya Raja
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Christopher Wu
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Esen Yonca Bassoy
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Thomas E Rubino
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Emma C Utagawa
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Paul M Magtibay
- Department of Gynecology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Kristina A Butler
- Department of Gynecology, Mayo Clinic, Scottsdale, Arizona, USA,College of Medicine and Science, Mayo Clinic, Scottsdale, Arizona, USA
| | - Marion Curtis
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA,College of Medicine and Science, Mayo Clinic, Scottsdale, Arizona, USA,Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| |
Collapse
|
195
|
Leveraging Natural Killer Cell Innate Immunity against Hematologic Malignancies: From Stem Cell Transplant to Adoptive Transfer and Beyond. Int J Mol Sci 2022; 24:ijms24010204. [PMID: 36613644 PMCID: PMC9820370 DOI: 10.3390/ijms24010204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Numerous recent advancements in T-cell based immunotherapies have revolutionized the treatment of hematologic malignancies. In the race towards the first approved allogeneic cellular therapy product, there is growing interest in utilizing natural killer (NK) cells as a platform for off-the-shelf cellular therapies due to their scalable manufacturing potential, potent anti-tumor efficacy, and superior safety profile. Allogeneic NK cell therapies are now being actively explored in the setting of hematopoietic stem cell transplantation and adoptive transfer. Increasingly sophisticated gene editing techniques have permitted the engineering of chimeric antigen receptors, ectopic cytokine expression, and tumor recognition signals to improve the overall cytotoxicity of NK cell therapies. Furthermore, the enhancement of antibody-dependent cellular cytotoxicity has been achieved through the use of NK cell engagers and combination regimens with monoclonal antibodies that act synergistically with CD16-expressing NK cells. Finally, a greater understanding of NK cell biology and the mechanisms of resistance have allowed the preclinical development of NK checkpoint blockade and methods to modulate the tumor microenvironment, which have been evaluated in early phase trials. This review will discuss the recent clinical advancements in NK cell therapies in hematologic malignancies as well as promising avenues of future research.
Collapse
|
196
|
Jin X, Bi J. Prospects for NK-based immunotherapy of chronic HBV infection. Front Immunol 2022; 13:1084109. [PMID: 36591230 PMCID: PMC9797727 DOI: 10.3389/fimmu.2022.1084109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/01/2022] [Indexed: 12/16/2022] Open
Abstract
Effective and long-term treatment is required for controlling chronic Hepatitis B Virus (HBV) infection. Natural killer (NK) cells are antiviral innate lymphocytes and represent an essential arm of current immunotherapy. In chronic HBV (CHB), NK cells display altered changes in phenotypes and functions, but preserve antiviral activity, especially for cytolytic activity. On the other hand, NK cells might also cause liver injury in the disease. NK -based immunotherapy, including adoptive NK cell therapy and NK -based checkpoint inhibition, could potentially exploit the antiviral aspect of NK cells for controlling CHB infection while preventing liver tissue damage. Here, we review recent progress in NK cell biology under the context of CHB infection, and discuss potential NK -based immunotherapy strategies for the disease.
Collapse
|
197
|
Friedmann KS, Kaschek L, Knörck A, Cappello S, Lünsmann N, Küchler N, Hoxha C, Schäfer G, Iden S, Bogeski I, Kummerow C, Schwarz EC, Hoth M. Interdependence of sequential cytotoxic T lymphocyte and natural killer cell cytotoxicity against melanoma cells. J Physiol 2022; 600:5027-5054. [PMID: 36226443 DOI: 10.1113/jp283667] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/06/2022] [Indexed: 01/05/2023] Open
Abstract
Cytotoxic T lymphocytes (CTL) and natural killer (NK) cells recognize and eliminate cancer cells. However, immune evasion, downregulation of immune function by the tumour microenvironment and resistance of cancer cells are major problems. Although CTL and NK cells are both important to eliminate cancer, most studies address them individually. We quantified sequential primary human CTL and NK cell cytotoxicity against the melanoma cell line SK-Mel-5. At high effector-to-target ratios, NK cells or melan-A (MART-1)-specific CTL eliminated all SK-Mel-5 cells within 24 h, indicating that SK-Mel-5 cells are not resistant initially. However, at lower effector-to-target ratios, which resemble numbers of the immune contexture in human cancer, a substantial number of SK-Mel-5 cells survived. Pre-exposure to CTL induced resistance in surviving SK-Mel-5 cells to subsequent CTL or NK cell cytotoxicity, and pre-exposure to NK cells induced resistance in surviving SK-Mel-5 cells to NK cells. Higher human leucocyte antigen class I expression or interleukin-6 levels were correlated with resistance to NK cells, whereas reduction in MART-1 antigen expression was correlated with reduced CTL cytotoxicity. The CTL cytotoxicity was rescued beyond control levels by exogenous MART-1 antigen. In contrast to the other three combinations, CTL cytotoxicity against SK-Mel-5 cells was enhanced following NK cell pre-exposure. Our assay allows quantification of sequential CTL and NK cell cytotoxicity and might guide strategies for efficient CTL-NK cell anti-melanoma therapies. KEY POINTS: Cytotoxic T lymphocytes (CTL) and natural killer (NK) cells eliminate cancer cells. Both CTL and NK cells attack the same targets, but most studies address them individually. In a sequential cytotoxicity model, the interdependence of antigen-specific CTL and NK cell cytotoxicity against melanoma is quantified. High numbers of antigen-specific CTL and NK cells eliminate all melanoma cells. However, lower numbers induce resistance if secondary CTL or NK cell exposure follows initial CTL exposure or if secondary NK cell exposure follows initial NK cell exposure. On the contrary, if secondary CTL exposure follows initial NK cell exposure, cytotoxicity is enhanced. Alterations in human leucocyte antigen class I expression and interleukin-6 levels are correlated with resistance to NK cells, whereas a reduction in antigen expression is correlated with reduced CTL cytotoxicity; CTL cytotoxicity is rescued beyond control levels by exogenous antigen. This assay and the results on interdependencies will help us to understand and optimize immune therapies against cancer.
Collapse
Affiliation(s)
- Kim S Friedmann
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Lea Kaschek
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Arne Knörck
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Sabrina Cappello
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany.,Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg August University, Göttingen, Germany
| | - Niklas Lünsmann
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Nadja Küchler
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Cora Hoxha
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Gertrud Schäfer
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Sandra Iden
- Cell and Developmental Biology, Center of Human and Molecular Biology (ZHMB), School of Medicine, Saarland University, Homburg, Germany
| | - Ivan Bogeski
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany.,Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg August University, Göttingen, Germany
| | - Carsten Kummerow
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
198
|
Modern Advances in CARs Therapy and Creating a New Approach to Future Treatment. Int J Mol Sci 2022; 23:ijms232315006. [PMID: 36499331 PMCID: PMC9739283 DOI: 10.3390/ijms232315006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Genetically engineered T and NK cells expressing a chimeric antigen receptor (CAR) are promising cytotoxic cells for the treatment of hematological malignancies and solid tumors. Despite the successful therapies using CAR-T cells, they have some disadvantages, such as cytokine release syndrome (CRS), neurotoxicity, or graft-versus-host-disease (GVHD). CAR-NK cells have lack or minimal cytokine release syndrome and neurotoxicity, but also multiple mechanisms of cytotoxic activity. NK cells are suitable for developing an "off the shelf" therapeutic product that causes little or no graft versus host disease (GvHD), but they are more sensitive to apoptosis and have low levels of gene expression compared to CAR-T cells. To avoid these adverse effects, further developments need to be considered to enhance the effectiveness of adoptive cellular immunotherapy. A promising approach to enhance the effectiveness of adoptive cellular immunotherapy is overcoming terminal differentiation or senescence and exhaustion of T cells. In this case, EVs derived from immune cells in combination therapy with drugs may be considered in the treatment of cancer patients, especially effector T and NK cells-derived exosomes with the cytotoxic activity of their original cells.
Collapse
|
199
|
Yu WF, Wang XQ, Zhao LP, Zhou JY, Feng JH. Down-regulation of IL-32γ expression reduces killing effect of natural killer cells on esophageal carcinoma cells. Shijie Huaren Xiaohua Zazhi 2022; 30:990-996. [DOI: 10.11569/wcjd.v30.i22.990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Interleukin (IL)-32γ is highly expressed in activated natural killer (NK) cells in esophageal cancer. However, it is not clear whether the expression level of IL-32γ in NK cells affects their killing effect on esophageal cancer cells.
AIM To investigate the role of IL-32γ in the anti-tumor effect of NK cells in esophageal cancer.
METHODS After transfecting NK-92 cells with shRNA targeting IL-32γ (shIL-32γ), the NK-92 cells were co-cultured with esophageal cancer cells EC9706 and TE-1, respectively. EC9706 and TE-1 cells were then collected; cell viability was measured by cell counting kit-8 (CCK-8) assay, cell proliferation was detected by 5-ethynyl-2'-deoxyuridine (EDU) assay, cell apoptosis was detected by flow cytometry, and the expression of apoptosis-related proteins B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X (Bax), cleaved cysteine-containing aspartate-specific proteases 3 (caspase 3), tumor necrosis factor receptor superfamily member 6 (FAS), death receptor 3 (DR3), and tumor necrosis factor receptor 2 (TNFR2) was detected by Western blot.
RESULTS After IL-32γ deletion in NK-92 cells, the cell viability and the EDU positive cells in EC9706 and TE-1 cells in the co-culture system were increased (P < 0.01), the expression level of Bcl-2 was increased (P < 0.01), and the expression levels of Bax, cleaved-caspase 3, FAS, DR3, and TNFR2 were all decreased (P < 0.01).
CONCLUSION Knockdown of IL-32γ attenuates the anti-tumor effect of NK-92 cells, which may be related to the inhibition of death receptor expression and caspase-3 activation in esophageal cancer cells.
Collapse
Affiliation(s)
- Wei-Fei Yu
- Graduate School, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China,Department of Tumor Chemoradiotherapy, Lishui People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Xiao-Qiu Wang
- Graduate School, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Li-Ping Zhao
- Graduate School, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jue-Yi Zhou
- Graduate School, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Ji-Hong Feng
- Graduate School, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| |
Collapse
|
200
|
Bahmanyar M, Vakil MK, Al-Awsi GRL, Kouhpayeh SA, Mansoori Y, Mansoori B, Moravej A, Mazarzaei A, Ghasemian A. Anticancer traits of chimeric antigen receptors (CARs)-Natural Killer (NK) cells as novel approaches for melanoma treatment. BMC Cancer 2022; 22:1220. [PMID: 36434591 PMCID: PMC9701052 DOI: 10.1186/s12885-022-10320-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Owing to non-responsiveness of a high number of patients to the common melanoma therapies, seeking novel approaches seem as an unmet requirement. Chimeric antigen receptor (CAR) T cells were initially employed against recurrent or refractory B cell malignancies. However, advanced stages or pretreated patients have insufficient T cells (lymphopenia) amount for collection and clinical application. Additionally, this process is time-consuming and logistically cumbersome. Another limitation of this approach is toxicity and cytokine release syndrome (CRS) progress and neurotoxicity syndrome (NS). Natural killer (NK) cells are a versatile component of the innate immunity and have several advantages over T cells in the application for therapies such as availability, unique biological features, safety profile, cost effectiveness and higher tissue residence. Additionally, CAR NK cells do not develop Graft-versus-host disease (GvHD) and are independent of host HLA genotype. Notably, the NK cells number and activity is affected in the tumor microenvironment (TME), paving the way for developing novel approaches by enhancing their maturation and functionality. The CAR NK cells short lifespan is a double edge sword declining toxicity and reducing their persistence. Bispecific and Trispecific Killer Cell Engagers (BiKE and Trike, respectively) are emerging and promising immunotherapies for efficient antibody dependent cell cytotoxicity (ADCC). CAR NK cells have some limitations in terms of expanding and transducing NK cells from donors to achieve clinical response. Clinical trials are in scarcity regarding the CAR NK cell-based cancer therapies. The CAR NK cells short life span following irradiation before infusion limits their efficiency inhibiting their in vivo expansion. The CAR NK cells efficacy enhancement in terms of lifespan TME preparation and stability is a goal for melanoma treatment. Combination therapies using CAR NK cells and chemotherapy can also overcome therapy limitations.
Collapse
Affiliation(s)
- Maryam Bahmanyar
- grid.411135.30000 0004 0415 3047Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mohammad Kazem Vakil
- grid.411135.30000 0004 0415 3047Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Seyed Amin Kouhpayeh
- grid.411135.30000 0004 0415 3047Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- grid.411135.30000 0004 0415 3047Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnam Mansoori
- grid.411135.30000 0004 0415 3047Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Moravej
- grid.411135.30000 0004 0415 3047Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdulbaset Mazarzaei
- grid.512728.b0000 0004 5907 6819Department of Immunology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Abdolmajid Ghasemian
- grid.411135.30000 0004 0415 3047Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|